1
|
Yavuz M, Kahyaogullari BN, Demircan T. Anti-carcinogenic effects of arecaidine but-2-ynyl ester tosylate on breast cancer: proliferation inhibition and activation of apoptosis. Mol Biol Rep 2025; 52:278. [PMID: 40035899 DOI: 10.1007/s11033-025-10385-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Breast cancer (BC) is the most prevalent cancer among women globally and is notoriously difficult to treat due to its heterogeneous nature and the lack of an effective treatment. Muscarinic receptors (MRs), which serve as key regulators in the parasympathetic nervous system, exhibit significant regulatory functions in non-neural cells. Recent studies suggest that modulating MR activity can elicit anti-carcinogenic effects across various malignancies, stimulating interest in their oncological implications. To investigate this further, we explored the anti-carcinogenic effects of arecaidine but-2-ynyl ester tosylate (ABET), a potential M2 receptor activator, in BC cells using several cellular and molecular assays. METHODS AND RESULTS Molecular docking assays were employed to confirm the binding affinity of ABET to M2/M4 receptors. Subsequently, we evaluated the impact of ABET on cell viability, proliferation, clonogenicity, and migration in MDA-MB-231 and MCF-7 BC cell lines. Computational analysis revealed preferential binding of ABET to M2 and M4 receptors. In-vitro experiments demonstrated that ABET markedly inhibits viability, growth, clonogenicity, and migration in BC cells. Notably, ABET induced cell cycle arrest in MDA-MB-231 cells and promoted apoptotic cell death in MCF-7 cells. Furthermore, ABET downregulated key proliferation- and cell cycle-associated genes, including CCND1, CDK6, and MKI67. CONCLUSIONS Our findings underscore ABET as a promising therapeutic candidate for BC treatment, capable of suppressing cell growth, survival, and migration. Additional in-vivo studies are necessary to validate ABET's anti-neoplastic efficacy and evaluate its feasibility as novel therapeutic agent in BC management.
Collapse
Affiliation(s)
- Mervenur Yavuz
- Institute of Natural Sciences, Department of Molecular Biology and Genetics, Muğla Sıtkı Koçman University, Muğla, Turkey
| | | | - Turan Demircan
- Medical Biology Department, School of Medicine, Muğla Sıtkı Koçman University, Muğla, Turkey.
| |
Collapse
|
2
|
Guerriero C, Fanfarillo R, Mancini P, Sterbini V, Guarguaglini G, Sforna L, Michelucci A, Catacuzzeno L, Tata AM. M2 muscarinic receptors negatively modulate cell migration in human glioblastoma cells. Neurochem Int 2024; 174:105673. [PMID: 38185384 DOI: 10.1016/j.neuint.2023.105673] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/17/2023] [Accepted: 12/29/2023] [Indexed: 01/09/2024]
Abstract
Glioblastoma (GB) is a very aggressive human brain tumor. The high growth potential and invasiveness make this tumor surgically and pharmacologically untreatable. Our previous work demonstrated that the activation of the M2 muscarinic acetylcholine receptors (M2 mAChRs) inhibited cell proliferation and survival in GB cell lines and in the cancer stem cells derived from human biopsies. The aim of the present study was to investigate the ability of M2 mAChR to modulate cell migration in two different GB cell lines: U87 and U251. By wound healing assay and single cell migration analysis performed by time-lapse microscopy, we demonstrated the ability of M2 mAChRs to negatively modulate cell migration in U251 but not in the U87 cell line. In order to explain the different effects observed in the two cell lines we have evaluated the possible involvement of the intermediate conductance calcium-activated potassium (IKCa) channel. IKCa channel is present in the GB cells, and it has been demonstrated to modulate cell migration. Using the perforated patch-clamp technique we have found that selective activation of M2 mAChR significantly reduced functional density of the IKCa current in U251 but not in U87 cells. To understand whether the M2 mAChR mediated reduction of ion channel density in the U251 cell line was relevant for the cell migration impairment, we tested the effects of TRAM-34, a selective inhibitor of the IKCa channel, in wound healing assay. We found that it was able to markedly reduce U251 cell migration and significantly decrease the number of invadopodia-like structure formations. These results suggest that only in U251 cells the reduced cell migration M2 mAChR-mediated might involve, at least in part, the IKCa channel.
Collapse
Affiliation(s)
- Claudia Guerriero
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185, Rome, Italy.
| | - Rachele Fanfarillo
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185, Rome, Italy.
| | - Patrizia Mancini
- Department Experimental Medicine, Sapienza University of Rome, 00185, Rome, Italy.
| | | | | | - Luigi Sforna
- Department of Chemistry Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - Antonio Michelucci
- Department of Chemistry Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - Luigi Catacuzzeno
- Department of Chemistry Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185, Rome, Italy; Research Centre of Neurobiology Daniel Bovet, Sapienza University of Rome, 00185, Rome, Italy.
| |
Collapse
|
3
|
Guerriero C, Manfredelli M, Matera C, Iuzzolino A, Conti L, Dallanoce C, De Amici M, Trisciuoglio D, Tata AM. M2 Muscarinic Receptor Stimulation Induces Autophagy in Human Glioblastoma Cancer Stem Cells via mTOR Complex-1 Inhibition. Cancers (Basel) 2023; 16:25. [PMID: 38201453 PMCID: PMC10778261 DOI: 10.3390/cancers16010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Although autophagy is a pro-survival process of tumor cells, it can stimulate cell death in particular conditions and when differently regulated by specific signals. We previously demonstrated that the selective stimulation of the M2 muscarinic receptor subtype (mAChR) negatively controls cell proliferation and survival and causes oxidative stress and cytotoxic and genotoxic effects in both GBM cell lines and GBM stem cells (GSCs). In this work, we have evaluated whether autophagy was induced as a downstream mechanism of the observed cytotoxic processes induced by M2 mAChR activation by the orthosteric agonist APE or the dualsteric agonist N8-Iper (N8). METHODS To assess the activation of autophagy, we analyzed the expression of LC3B using Western blot analysis and in LC3B-EGFP transfected cell lines. Apoptosis was assessed by measuring the protein expression of Caspases 3 and 9. RESULTS Our data indicate that activation of M2 mAChR by N8 promotes autophagy in both U251 and GB7 cell lines as suggested by the LC3B-II expression level and analysis of the transfected cells by fluorescence microscopy. Autophagy induction by M2 mAChRs is regulated by the decreased activity of the PI3K/AKT/mTORC1 pathway and upregulated by pAMPK expression. Downstream of autophagy activation, an increase in apoptosis was also observed in both cell lines after treatment with the two M2 agonists. CONCLUSIONS N8 treatment causes autophagy via pAMPK upregulation, followed by apoptosis in both investigated cell lines. In contrast, the absence of autophagy in APE-treated GSC cells seems to indicate that cell death could be triggered by mechanisms alternative to those observed for N8.
Collapse
Affiliation(s)
- Claudia Guerriero
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (M.M.); (A.I.)
| | - Marianna Manfredelli
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (M.M.); (A.I.)
| | - Carlo Matera
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; (C.M.); (C.D.); (M.D.A.)
| | - Angela Iuzzolino
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (M.M.); (A.I.)
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy;
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology—CIBIO, University of Trento, 38123 Trento, Italy;
| | - Clelia Dallanoce
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; (C.M.); (C.D.); (M.D.A.)
| | - Marco De Amici
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; (C.M.); (C.D.); (M.D.A.)
| | - Daniela Trisciuoglio
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy;
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (M.M.); (A.I.)
- Research Centre of Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy
- Consortium Interuniversity Biotechnologies (CIB), University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
4
|
Zhou X, Wang X, Li N, Guo Y, Yang X, Lei Y. Therapy resistance in neuroblastoma: Mechanisms and reversal strategies. Front Pharmacol 2023; 14:1114295. [PMID: 36874032 PMCID: PMC9978534 DOI: 10.3389/fphar.2023.1114295] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Neuroblastoma is one of the most common pediatric solid tumors that threaten the health of children, accounting for about 15% of childhood cancer-related mortality in the United States. Currently, multiple therapies have been developed and applied in clinic to treat neuroblastoma including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. However, the resistance to therapies is inevitable following long-term treatment, leading to treatment failure and cancer relapse. Hence, to understand the mechanisms of therapy resistance and discover reversal strategies have become an urgent task. Recent studies have demonstrated numerous genetic alterations and dysfunctional pathways related to neuroblastoma resistance. These molecular signatures may be potential targets to combat refractory neuroblastoma. A number of novel interventions for neuroblastoma patients have been developed based on these targets. In this review, we focus on the complicated mechanisms of therapy resistance and the potential targets such as ATP-binding cassette transporters, long non-coding RNAs, microRNAs, autophagy, cancer stem cells, and extracellular vesicles. On this basis, we summarized recent studies on the reversal strategies to overcome therapy resistance of neuroblastoma such as targeting ATP-binding cassette transporters, MYCN gene, cancer stem cells, hypoxia, and autophagy. This review aims to provide novel insight in how to improve the therapy efficacy against resistant neuroblastoma, which may shed light on the future directions that would enhance the treatment outcomes and prolong the survival of patients with neuroblastoma.
Collapse
Affiliation(s)
- Xia Zhou
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiaokang Wang
- Department of Pharmacy, Shenzhen Longhua District Central Hospital, Shenzhen, China.,Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Nan Li
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yu Guo
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaolin Yang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuhe Lei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
5
|
Taggi M, Kovacevic A, Capponi C, Falcinelli M, Cacciamani V, Vicini E, Canipari R, Tata AM. The activation of M2 muscarinic receptor inhibits cell growth and survival in human epithelial ovarian carcinoma. J Cell Biochem 2022; 123:1440-1453. [PMID: 35775813 DOI: 10.1002/jcb.30303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 01/23/2023]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related deaths in females. Many ovarian tumor cell lines express muscarinic receptors (mAChRs), and their expression is correlated with reduced survival of patients. We have characterized the expression of mAChRs in two human ovarian carcinoma cell lines (SKOV-3, TOV-21G) and two immortalized ovarian surface epithelium cell lines (iOSE-120, iOSE-398). Among the five subtypes of mAChRs (M1-M5 receptors), we focused our attention on the M2 receptor, which is involved in the inhibition of tumor cell proliferation. Western blot analysis and real-time PCR analyses indicated that the levels of M2 are statistically downregulated in cancer cells. Therefore, we investigated the effect of arecaidine propargyl ester hydrobromide (APE), a preferential M2 agonist, on cell growth and survival. APE treatment decreased cell number in a dose and time-dependent manner by decreasing cell proliferation and increasing cell death. FACS and immunocytochemistry analysis have also demonstrated the ability of APE to accumulate the cells in G2/M phase of the cell cycle and to increase the percentage of abnormal mitosis. The higher level of M2 receptors in the iOSE cells rendered these cells more sensitive to APE treatment than cancer cells. The data here reported suggest that M2 has a negative role in cell growth/survival of ovarian cell lines, and its downregulation may favor tumor progression.
Collapse
Affiliation(s)
- Marilena Taggi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Andjela Kovacevic
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Chiara Capponi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Marta Falcinelli
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Veronica Cacciamani
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Elena Vicini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Rita Canipari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Analysis of Signal Transduction Pathways Downstream M2 Receptor Activation: Effects on Schwann Cell Migration and Morphology. Life (Basel) 2022; 12:life12020211. [PMID: 35207498 PMCID: PMC8875146 DOI: 10.3390/life12020211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/14/2023] Open
Abstract
Background: Schwann cells (SCs) express cholinergic receptors, suggesting a role of cholinergic signaling in the control of SC proliferation, differentiation and/or myelination. Our previous studies largely demonstrated that the pharmacological activation of the M2 muscarinic receptor subtype caused an inhibition of cell proliferation and promoted the expression of pro-myelinating differentiation genes. In order to elucidate the molecular signaling activated downstream the M2 receptor activation, in the present study we investigated the signal transduction pathways activated by the M2 orthosteric agonist arecaidine propargyl ester (APE) in SCs. Methods: Using Western blot we analyzed some components of the noncanonical pathways involving β1-arrestin and PI3K/AKT/mTORC1 signaling. A wound healing assay was used to evaluate SC migration. Results: Our results demonstrated that M2 receptor activation negatively modulated the PI3K/Akt/mTORC1 axis, possibly through β1-arrestin downregulation. The involvement of the mTORC1 complex was also supported by the decreased expression of its specific target p-p70 S6KThr389. Then, we also analyzed the expression of p-AMPKαthr172, a negative regulator of myelination that resulted in reduced levels after M2 agonist treatment. The analysis of cell migration and morphology allowed us to demonstrate that M2 receptor activation caused an arrest of SC migration and modified cell morphology probably by the modulation of β1-arrestin/cofilin-1 and PKCα expression, respectively. Conclusions: The data obtained demonstrated that M2 receptor activation in addition to the canonical Gi protein-coupled pathway modulates noncanonical pathways involving the mTORC1 complex and other kinases whose activation may contribute to the inhibition of SC proliferation and migration and address SC differentiation.
Collapse
|
7
|
Zhang L, Wang M, Zhu Z, Chen S, Wu H, Yang Y, Che F, Li Q, Li H. A GD2-aptamer-mediated, self-assembling nanomedicine for targeted multiple treatments in neuroblastoma theranostics. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:732-748. [PMID: 34703655 PMCID: PMC8515170 DOI: 10.1016/j.omtn.2021.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
Because current mainstream anti-glycolipid GD2 therapeutics for neuroblastoma (NB) have limitations, such as severe adverse effects, improved therapeutics are needed. In this study, we developed a GD2 aptamer (DB99) and constructed a GD2-aptamer-mediated multifunctional nanomedicine (ANM) with effective, precise, and biocompatible properties, which functioned both as chemotherapy and as gene therapy for NB. DB99 can bind to GD2+ NB tumor cells but has minimal cross-reactivity to GD2− cells. Furthermore, ANM is formulated by self-assembly of synthetic aptamers DB99 and NB-specific MYCN small interfering RNA (siRNA), followed by self-loading of the chemotherapeutic agent doxorubicin (Dox). ANM is capable of specifically recognizing, binding, and internalizing GD2+, but not GD2−, NB tumor cells in vitro. Intracellular delivery of ANM activates Dox release for chemotherapy and MYCN-siRNA-induced MYCN silencing. ANM specifically targets, and selectively accumulates in, the GD2+ tumor site in vivo and further induces growth inhibition of GD2+ tumors in vivo; in addition, ANM generates fewer or no side effects in healthy tissues, resulting in markedly longer survival with fewer adverse effects. These results suggest that the GD2-aptamer-mediated, targeted drug delivery system may have potential applications for precise treatment of NB.
Collapse
Affiliation(s)
- Liyu Zhang
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.,Shaanxi Institute of Pediatric Diseases, Affiliated Children's hospital of Xi'an Jiaotong University, Xi'an 710002, Shaanixi, China
| | - Meng Wang
- Department of Emergency Surgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Zeen Zhu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Shengquan Chen
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Haibin Wu
- Shaanxi Institute of Pediatric Diseases, Affiliated Children's hospital of Xi'an Jiaotong University, Xi'an 710002, Shaanixi, China
| | - Ying Yang
- Shaanxi Institute of Pediatric Diseases, Affiliated Children's hospital of Xi'an Jiaotong University, Xi'an 710002, Shaanixi, China
| | - Fengyu Che
- Shaanxi Institute of Pediatric Diseases, Affiliated Children's hospital of Xi'an Jiaotong University, Xi'an 710002, Shaanixi, China
| | - Qiao Li
- Department of clinical laboratory, Affiliated Children's hospital of Xi'an Jiaotong University, Xi'an 710002, China
| | - Hui Li
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.,Department of Neonatology, Affiliated Children's hospital of Xi'an Jiaotong University, Xi'an 710002, China
| |
Collapse
|
8
|
Guerriero C, Matera C, Del Bufalo D, De Amici M, Conti L, Dallanoce C, Tata AM. The Combined Treatment with Chemotherapeutic Agents and the Dualsteric Muscarinic Agonist Iper-8-Naphthalimide Affects Drug Resistance in Glioblastoma Stem Cells. Cells 2021; 10:cells10081877. [PMID: 34440646 PMCID: PMC8391681 DOI: 10.3390/cells10081877] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is characterized by heterogeneous cell populations. Among these, the Glioblastoma Stem Cells (GSCs) fraction shares some similarities with Neural Stem Cells. GSCs exhibit enhanced resistance to conventional chemotherapy drugs. Our previous studies demonstrated that the activation of M2 muscarinic acetylcholine receptors (mAChRs) negatively modulates GSCs proliferation and survival. The aim of the present study was to analyze the ability of the M2 dualsteric agonist Iper-8-naphthalimide (N-8-Iper) to counteract GSCs drug resistance. METHODS Chemosensitivity to M2 dualsteric agonist N-8-Iper and chemotherapy drugs such as temozolomide, doxorubicin, or cisplatin was evaluated in vitro by MTT assay in two different GSC lines. Drug efflux pumps expression was evaluated by RT-PCR and qRT-PCR. RESULTS By using sub-toxic concentrations of N-8-Iper combined with the individual chemotherapeutic agents, we found that only low doses of the M2 agonist combined with doxorubicin or cisplatin or temozolomide were significantly able to counteract cell growth in both GSC lines. Moreover, we evaluated as the exposure to high and low doses of N-8-Iper downregulated the ATP-binding cassette (ABC) drug efflux pumps expression levels. CONCLUSIONS Our results revealed the ability of the investigated M2 agonist to counteract drug resistance in two GSC lines, at least partially by downregulating the ABC drug efflux pumps expression. The combined effects of low doses of conventional chemotherapy and M2 agonists may thus represent a novel promising pharmacological approach to impair the GSC-drug resistance in the GBM therapy.
Collapse
Affiliation(s)
- Claudia Guerriero
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy;
| | - Carlo Matera
- Department of Pharmaceutical Sciences, Medicinal Chemistry Section “Pietro Pratesi”, University of Milan, 20133 Milan, Italy; (C.M.); (M.D.A.); (C.D.)
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, Regina Elena National Cancer Institute, 00187 Rome, Italy;
| | - Marco De Amici
- Department of Pharmaceutical Sciences, Medicinal Chemistry Section “Pietro Pratesi”, University of Milan, 20133 Milan, Italy; (C.M.); (M.D.A.); (C.D.)
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology—CIBIO, University of Trento, 38123 Trento, Italy;
| | - Clelia Dallanoce
- Department of Pharmaceutical Sciences, Medicinal Chemistry Section “Pietro Pratesi”, University of Milan, 20133 Milan, Italy; (C.M.); (M.D.A.); (C.D.)
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy;
- Research Centre of Neurobiology Daniel Bovet, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
9
|
Di Bari M, Tombolillo V, Alessandrini F, Guerriero C, Fiore M, Asteriti IA, Castigli E, Sciaccaluga M, Guarguaglini G, Degrassi F, Tata AM. M2 Muscarinic Receptor Activation Impairs Mitotic Progression and Bipolar Mitotic Spindle Formation in Human Glioblastoma Cell Lines. Cells 2021; 10:cells10071727. [PMID: 34359896 PMCID: PMC8306299 DOI: 10.3390/cells10071727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/24/2021] [Accepted: 07/06/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Glioblastoma multiforme (GBM) is characterized by several genetic abnormalities, leading to cell cycle deregulation and abnormal mitosis caused by a defective checkpoint. We previously demonstrated that arecaidine propargyl ester (APE), an orthosteric agonist of M2 muscarinic acetylcholine receptors (mAChRs), arrests the cell cycle of glioblastoma (GB) cells, reducing their survival. The aim of this work was to better characterize the molecular mechanisms responsible for this cell cycle arrest. Methods: The arrest of cell proliferation was evaluated by flow cytometry analysis. Using immunocytochemistry and time-lapse analysis, the percentage of abnormal mitosis and aberrant mitotic spindles were assessed in both cell lines. Western blot analysis was used to evaluate the modulation of Sirtuin2 and acetylated tubulin—factors involved in the control of cell cycle progression. Results: APE treatment caused arrest in the M phase, as indicated by the increase in p-HH3 (ser10)-positive cells. By immunocytochemistry, we found a significant increase in abnormal mitoses and multipolar mitotic spindle formation after APE treatment. Time-lapse analysis confirmed that the APE-treated GB cells were unable to correctly complete the mitosis. The modulated expression of SIRT2 and acetylated tubulin in APE-treated cells provides new insights into the mechanisms of altered mitotic progression in both GB cell lines. Conclusions: Our data show that the M2 agonist increases aberrant mitosis in GB cell lines. These results strengthen the idea of considering M2 acetylcholine receptors a novel promising therapeutic target for the glioblastoma treatment.
Collapse
Affiliation(s)
- Maria Di Bari
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (M.D.B.); (V.T.); (F.A.); (C.G.)
| | - Vanessa Tombolillo
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (M.D.B.); (V.T.); (F.A.); (C.G.)
| | - Francesco Alessandrini
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (M.D.B.); (V.T.); (F.A.); (C.G.)
| | - Claudia Guerriero
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (M.D.B.); (V.T.); (F.A.); (C.G.)
| | - Mario Fiore
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (M.F.); (I.A.A.); (G.G.); (F.D.)
| | - Italia Anna Asteriti
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (M.F.); (I.A.A.); (G.G.); (F.D.)
| | - Emilia Castigli
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, 06100 Perugia, Italy;
| | - Miriam Sciaccaluga
- Department of Medicine and Surgery, University of Perugia, 06100 Perugia, Italy;
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (M.F.); (I.A.A.); (G.G.); (F.D.)
| | - Francesca Degrassi
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (M.F.); (I.A.A.); (G.G.); (F.D.)
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (M.D.B.); (V.T.); (F.A.); (C.G.)
- Research Centre of Neurobiology Daniel Bovet, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
10
|
Español A, Salem A, Sanchez Y, Sales ME. Breast cancer: Muscarinic receptors as new targets for tumor therapy. World J Clin Oncol 2021; 12:404-428. [PMID: 34189066 PMCID: PMC8223712 DOI: 10.5306/wjco.v12.i6.404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
The development of breast cancer is a complex process that involves the participation of different factors. Several authors have demonstrated the overexpression of muscarinic acetylcholine receptors (mAChRs) in different tumor tissues and their role in the modulation of tumor biology, positioning them as therapeutic targets in cancer. The conventional treatment for breast cancer involves surgery, radiotherapy, and/or chemotherapy. The latter presents disadvantages such as limited specificity, the appearance of resistance to treatment and other side effects. To prevent these side effects, several schedules of drug administration, like metronomic therapy, have been developed. Metronomic therapy is a type of chemotherapy in which one or more drugs are administered at low concentrations repetitively. Recently, two chemotherapeutic agents usually used to treat breast cancer have been considered able to activate mAChRs. The combination of low concentrations of these chemotherapeutic agents with muscarinic agonists could be a useful option to be applied in breast cancer treatment, since this combination not only reduces tumor cell survival without affecting normal cells, but also decreases pathological neo-angiogenesis, the expression of drug extrusion proteins and the cancer stem cell fraction. In this review, we focus on the previous evidences that have positioned mAChRs as relevant therapeutic targets in breast cancer and analyze the effects of administering muscarinic agonists in combination with conventional chemotherapeutic agents in a metronomic schedule.
Collapse
Affiliation(s)
- Alejandro Español
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Agustina Salem
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Yamila Sanchez
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - María Elena Sales
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| |
Collapse
|